Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
EMBO Rep ; 18(7): 1150-1165, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507162

RESUMEN

Dihydroceramide desaturases are evolutionarily conserved enzymes that convert dihydroceramide (dhCer) to ceramide (Cer). While elevated Cer levels cause neurodegenerative diseases, the neuronal activity of its direct precursor, dhCer, remains unclear. We show that knockout of the fly dhCer desaturase gene, infertile crescent (ifc), results in larval lethality with increased dhCer and decreased Cer levels. Light stimulation leads to ROS increase and apoptotic cell death in ifc-KO photoreceptors, resulting in activity-dependent neurodegeneration. Lipid-containing Atg8/LC3-positive puncta accumulate in ifc-KO photoreceptors, suggesting lipophagy activation. Further enhancing lipophagy reduces lipid droplet accumulation and rescues ifc-KO defects, indicating that lipophagy plays a protective role. Reducing dhCer synthesis prevents photoreceptor degeneration and rescues ifc-KO lethality, while supplementing downstream sphingolipids does not. These results pinpoint that dhCer accumulation is responsible for ifc-KO defects. Human dhCer desaturase rescues ifc-KO larval lethality, and rapamycin reverses defects caused by dhCer accumulation in human neuroblastoma cells, suggesting evolutionarily conserved functions. This study demonstrates a novel requirement for dhCer desaturase in neuronal maintenance in vivo and shows that lipophagy activation prevents activity-dependent degeneration caused by dhCer accumulation.


Asunto(s)
Autofagia , Ceramidas/metabolismo , Metabolismo de los Lípidos , Animales , Apoptosis , Línea Celular Tumoral , Ceramidas/análisis , Drosophila , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Ácido Graso Desaturasas/genética , Técnicas de Inactivación de Genes , Humanos , Luz/efectos adversos , Lipólisis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Enfermedades Neurodegenerativas/prevención & control , Células Fotorreceptoras de Invertebrados/patología , Células Fotorreceptoras de Invertebrados/efectos de la radiación , Esfingolípidos/metabolismo
2.
Int J Mol Sci ; 18(8)2017 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-28825666

RESUMEN

Neuropeptide FF (NPFF) belongs to the RFamide family and is known as a morphine-modulating peptide. NPFF regulates various hypothalamic functions through two receptors, NPFFR1 and NPFFR2. The hypothalamic-pituitary-adrenal (HPA) axis participates in physiological stress response by increasing circulating glucocorticoid levels and modulating emotional responses. Other RFamide peptides, including neuropeptide AF, neuropeptide SF and RFamide related peptide also target NPFFR1 or NPFFR2, and have been reported to activate the HPA axis and induce anxiety- or depression-like behaviors. However, little is known about the action of NPFF on HPA axis activity and anxiety-like behaviors, and the role of the individual receptors remains unclear. In this study, NPFFR2 agonists were used to examine the role of NPFFR2 in activating the HPA axis in rodents. Administration of NPFFR2 agonists, dNPA (intracerebroventricular, ICV) and AC-263093 (intraperitoneal, IP), time-dependently (in rats) and dose-dependently (in mice) increased serum corticosteroid levels and the effects were counteracted by the NPFF receptor antagonist, RF9 (ICV), as well as corticotropin-releasing factor (CRF) antagonist, α-helical CRF(9-41) (intravenous, IV). Treatment with NPFFR2 agonist (AC-263093, IP) increased c-Fos protein expression in the hypothalamic paraventricular nucleus and induced an anxiogenic effect, which was evaluated in mice using an elevated plus maze. These findings reveal, for the first time, that the direct action of hypothalamic NPFFR2 stimulates the HPA axis and triggers anxiety-like behaviors.


Asunto(s)
Trastorno Depresivo/metabolismo , Oligopéptidos/metabolismo , Receptores de Neuropéptido/metabolismo , Corticoesteroides/sangre , Animales , Ansiedad , Hormona Liberadora de Corticotropina/administración & dosificación , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Trastorno Depresivo/sangre , Trastorno Depresivo/fisiopatología , Hidrazinas/administración & dosificación , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/metabolismo , Hipotálamo/patología , Ratones , Oligopéptidos/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Ratas , Receptores de Neuropéptido/agonistas
3.
Cell Rep ; 35(2): 108972, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852856

RESUMEN

Disruption of sphingolipid homeostasis is known to cause neurological disorders, but the mechanisms by which specific sphingolipid species modulate pathogenesis remain unclear. The last step of de novo sphingolipid synthesis is the conversion of dihydroceramide to ceramide by dihydroceramide desaturase (human DEGS1; Drosophila Ifc). Loss of ifc leads to dihydroceramide accumulation, oxidative stress, and photoreceptor degeneration, whereas human DEGS1 variants are associated with leukodystrophy and neuropathy. In this work, we demonstrate that DEGS1/ifc regulates Rac1 compartmentalization in neuronal cells and that dihydroceramide alters the association of active Rac1 with organelle-mimicking membranes. We further identify the Rac1-NADPH oxidase (NOX) complex as the major cause of reactive oxygen species (ROS) accumulation in ifc-knockout (ifc-KO) photoreceptors and in SH-SY5Y cells with the leukodystrophy-associated DEGS1H132R variant. Suppression of Rac1-NOX activity rescues degeneration of ifc-KO photoreceptors and ameliorates oxidative stress in DEGS1H132R-carrying cells. Therefore, we conclude that DEGS1/ifc deficiency causes dihydroceramide accumulation, resulting in Rac1 mislocalization and NOX-dependent neurodegeneration.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ácido Graso Desaturasas/genética , Proteínas de la Membrana/genética , NADPH Oxidasas/genética , Proteína de Unión al GTP rac1/genética , Animales , Línea Celular Tumoral , Ceramidas/metabolismo , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/metabolismo , Electrorretinografía , Ácido Graso Desaturasas/antagonistas & inhibidores , Ácido Graso Desaturasas/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/deficiencia , NADPH Oxidasas/metabolismo , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patología , Mutación Puntual , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retina/metabolismo , Retina/patología , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
4.
Psychoneuroendocrinology ; 71: 73-85, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27243477

RESUMEN

Neuropeptide FF (NPFF) is a morphine-modulating peptide that regulates the analgesic effect of opioids, and also controls food consumption and cardiovascular function through its interaction with two cognate receptors, NPFFR1 and NPFFR2. In the present study, we explore a novel modulatory role for NPFF-NPFFR2 in stress-related depressive behaviors. In a mouse model of chronic mild stress (CMS)-induced depression, the expression of NPFF significantly increased in the hypothalamus, hippocampus, medial prefrontal cortex (mPFC) and amygdala. In addition, transgenic (Tg) mice over-expressing NPFFR2 displayed clear depression and anxiety-like behaviors with hyperactivity in the hypothalamic-pituitary-adrenal (HPA) axis, reduced expression of glucocorticoid receptor (GR) and neurogenesis in the hippocampus. Furthermore, acute treatment of NPFFR2 agonists in wild-type (WT) mice enhanced the activity of the HPA axis, and chronic administration resulted in depressive and anxiety-like behaviors. Chronic stimulation of NPFFR2 also decreased the expression of hippocampal GR and led to persistent activation of the HPA axis. Strikingly, bilateral intra-paraventricular nucleus (PVN) injection of NPFFR2 shRNA predominately inhibits the depressive-like behavior in CMS-exposed mice. Antidepressants, fluoxetine and ketamine, effectively relieved the depressive behaviors of NPFFR2-Tg mice. We speculate that persistent NPFFR2 activation, in particular in the hypothalamus, up-regulates the HPA axis and results in long-lasting increases in circulating corticosterone (CORT), consequently damaging hippocampal function. This novel role of NPFFR2 in regulating the HPA axis and hippocampal function provides a new avenue for combating depression and anxiety-like disorder.


Asunto(s)
Depresión/fisiopatología , Receptores de Neuropéptido/metabolismo , Animales , Antidepresivos/farmacología , Ansiedad , Trastornos de Ansiedad/metabolismo , Enfermedad Crónica/psicología , Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Trastorno Depresivo/fisiopatología , Expresión Génica , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Corteza Prefrontal/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Neuropéptido/fisiología , Estrés Fisiológico/efectos de los fármacos , Estrés Psicológico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA